8
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Synthetic augmentation of bilirubin metabolism in human pluripotent stem cell-derived liver organoids

      brief-report

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Summary

          UGT1A1 (UDP glucuronosyltransferase family 1 member A1) is the primary enzyme required for bilirubin conjugation, which is essential for preventing hyperbilirubinemia. Animal models lack key human organic anion transporting polypeptides with distinct epigenetic control over bilirubin metabolism, necessitating a human model to interrogate the regulatory mechanism behind UGT1A1 function. Here, we use induced pluripotent stem cells to develop human liver organoids that can emulate conjugation failure phenotype. Bilirubin conjugation assays, chromatin immunoprecipitation, and transcriptome analysis elucidated the role of glucocorticoid antagonism in UGT1A1 activation. This antagonism prevents the binding of transcriptional repressor MECP2 at the expense of NRF2 with associated off-target effects. Therefore, we introduced functional GULO (L-gulonolactone oxidase) in human organoids to augment intracellular ascorbate for NRF2 reactivation. This engineered organoid conjugated more bilirubin and protected against hyperbilirubinemia when transplanted in immunosuppressed Crigler-Najjar syndrome rat model. Collectively, we demonstrate that our organoid system serves as a manipulatable model for interrogating hyperbilirubinemia and potential therapeutic development.

          Graphical abstract

          Highlights

          • HLOs are amenable for studying mediated bilirubin conjugation

          • HLOs inform the glucocorticoid-mediated epigenetic regulation of UGT1A1

          • Synthetic introduction of murine GULO in HLOs more efficiently conjugates bilirubin

          • Engineered HLO transplantation alleviates the symptoms of Crigler-Najjar syndrome model

          Abstract

          This paper describes the development of pluripotent stem cell-derived human liver organoid (HLO) models to study UGT1A1 gene regulation and function. We engineered active GULO (murine gulonolactone oxidase) expressing HLO to augment therapeutic potential, thus alleviating symptoms in Crigler-Najjar syndrome model rats upon transplantation.

          Related collections

          Most cited references45

          • Record: found
          • Abstract: found
          • Article: not found

          Modeling Steatohepatitis in Humans with Pluripotent Stem Cell-Derived Organoids

          Human organoid systems recapitulate in vivo organ architecture, yet fail to capture complex pathologies such as inflammation and fibrosis. Here, using 11 different healthy and diseased pluripotent stem cell lines, we developed a reproducible method to derive multi-cellular human liver organoids composed of hepatocyte-, stellate- and Kupffer-like cells that exhibit transcriptomic resemblance to in vivo derived tissues. Under free fatty acid treatment, organoids, but not reaggregated cocultured spheroids, recapitulated key features of steatohepatitis including steatosis, inflammation and fibrosis phenotypes in a successive manner. Interestingly, an organoid-level biophysical readout with atomic force microscopy demonstrated that organoid stiffening reflects the fibrosis severity. Furthermore, organoids from patients with genetic dysfunction of lysosomal acid lipase phenocopied severe steatohepatitis, rescued by FXR agonism-mediated reactive oxygen species suppression. The presented key methodology and preliminary results offer a new approach for studying personalized basis for inflammation and fibrosis in human, thus facilitating the discovery of effective treatments. Ouchi et al. develop a reproducible method to generate multi-cellular human liver organoids from iPSC/ESCs. The organoids recapitulate progressive features of steatohepatitis including steatosis, inflammation and fibrosis. A patient-derived organoid with lysosomal acid lipase deficiency exhibits the exaggerated steatohepatitis phenotype, as seen in vivo , and can be rescued by FGF19.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            The Keap1–Nrf2 system in cancers: stress response and anabolic metabolism

            The Keap1–Nrf2 [Kelch-like ECH-associated protein 1–nuclear factor (erythroid-derived 2)-like 2] pathway plays a central role in the protection of cells against oxidative and xenobiotic stresses. Nrf2 is a potent transcription activator that recognizes a unique DNA sequence known as the antioxidant response element (ARE). Under normal conditions, Nrf2 binds to Keap1 in the cytoplasm, resulting in proteasomal degradation. Following exposure to electrophiles or reactive oxygen species, Nrf2 becomes stabilized, translocates into the nucleus, and activates the transcription of various cytoprotective genes. Increasing attention has been paid to the role of Nrf2 in cancer cells because the constitutive stabilization of Nrf2 has been observed in many human cancers with poor prognosis. Recent studies have shown that the antioxidant and detoxification activities of Nrf2 confer chemo- and radio-resistance to cancer cells. In this review, we provide an overview of the Keap1–Nrf2 system and discuss its role under physiological and pathological conditions, including cancers. We also introduce the results of our recent study describing Nrf2 function in the metabolism of cancer cells. Nrf2 likely confers a growth advantage to cancer cells through enhancing cytoprotection and anabolism. Finally, we discuss the possible impact of Nrf2 inhibitors on cancer therapy.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              A bilirubin-inducible fluorescent protein from eel muscle.

              The fluorescent protein toolbox has revolutionized experimental biology. Despite this advance, no fluorescent proteins have been identified from vertebrates, nor has chromogenic ligand-inducible activation or clinical utility been demonstrated. Here, we report the cloning and characterization of UnaG, a fluorescent protein from Japanese eel. UnaG belongs to the fatty-acid-binding protein (FABP) family, and expression in eel is restricted to small-diameter muscle fibers. On heterologous expression in cell lines or mouse brain, UnaG produces oxygen-independent green fluorescence. Remarkably, UnaG fluorescence is triggered by an endogenous ligand, bilirubin, a membrane-permeable heme metabolite and clinical health biomarker. The holoUnaG structure at 1.2 Å revealed a biplanar coordination of bilirubin by reversible π-conjugation, and we used this high-affinity and high-specificity interaction to establish a fluorescence-based human bilirubin assay with promising clinical utility. UnaG will be the prototype for a versatile class of ligand-activated fluorescent proteins, with applications in research, medicine, and bioengineering. Copyright © 2013 Elsevier Inc. All rights reserved.
                Bookmark

                Author and article information

                Contributors
                Journal
                Stem Cell Reports
                Stem Cell Reports
                Stem Cell Reports
                Elsevier
                2213-6711
                12 October 2023
                14 November 2023
                12 October 2023
                : 18
                : 11
                : 2071-2083
                Affiliations
                [1 ]Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
                [2 ]Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
                [3 ]Division of Gastroenterology, Hepatology & Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
                [4 ]College of Medicine, University of Kentucky, Lexington, KY, USA
                [5 ]Department of Transplantation Surgery, Nagoya University Hospital, Nagoya University Graduate School of Medicine, Aichi, Japan
                [6 ]Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
                [7 ]Institute of Research, Tokyo Medical and Dental University, Tokyo, Japan
                [8 ]Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
                [9 ]Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), and Division of Stem Cell and Organoid Medicine, Osaka University, Suita, Osaka 565-0871, Japan
                Author notes
                []Corresponding author takanori.takebe@ 123456cchmc.org
                Article
                S2213-6711(23)00365-X
                10.1016/j.stemcr.2023.09.006
                10679658
                37832542
                868abdf2-f92d-4c3d-a6dc-11219b9fb8ee
                © 2023 The Authors

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                : 24 January 2023
                : 13 September 2023
                : 14 September 2023
                Categories
                Report

                liver organoid,bilirubin conjugation,glucocorticoid signaling,l-gulonolactone oxidase,transplantation

                Comments

                Comment on this article